You are on page 1of 13

European Heart Journal (2023) 44, 643–655 STATE OF THE ART REVIEW

https://doi.org/10.1093/eurheartj/ehac759 Heart failure and cardiomyopathies

Endocrine functions of the heart: from bench


to bedside
1,2 1
Massimo Volpe *, Giovanna Gallo , and Speranza Rubattu1,3
1
Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy; 2IRCCS San Raffaele, Via della Pisana 235, 00163 Rome, Italy; and

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


3
IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli (IS), Italy

Received 11 July 2022; revised 22 November 2022; accepted 5 December 2022; online publish-ahead-of-print 30 December 2022

Graphical Abstract

FFA oxidation
GH secretion
Regulation of body growth

Natriuresis/diuresis
GLP-1
RAAS inhibition
Insulin secretion
Blood volume reduction

Endocrine
Endo
Endocrine ffunction
unction off tthe
he heart
he
earr t
Natriu
Na
Natriuretic
ri retic
etic peptides
tide
Other cardiac hormones
Vasorelaxation Oxygen consumption
BP reduction FFA oxidation
Increased capillary Regulation of muscle mass
permeability

Lipolysis
SNS inhibition
Postprandial energy expenditure
Reduced AVP secrection
Mitochondrial respiration

The graphical abstract illustrates the systemic effects of NPs and other hormones secreted from the heart, their peripheral effects, and the inter­
actions with other endocrine systems. AVP, arginine vasopressin; BP, blood pressure; FFA, free fatty acid; GH, growth hormone; GLP-1, glucagon like
peptide-1; RAAS, renin-angiotensin-aldosterone system; SNS, sympathetic nervous system.

* Corresponding author. Tel: +39 06 33775979, Fax: +39 06 33775546, Email: massimo.volpe@uniroma1.it
© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
644 M. Volpe et al.

Abstract

Heart has a recognized endocrine function as it produces several biologically active substances with hormonal properties. Among these hormones,
the natriuretic peptide (NP) system has been extensively characterized and represents a prominent expression of the endocrine function of the
heart. Over the years, knowledge about the mechanisms governing their synthesis, secretion, processing, and receptors interaction of NPs has
been intensively investigated. Their main physiological endocrine and paracrine effects on cardiovascular and renal systems are mostly mediated
through guanylate cyclase-A coupled receptors. The potential role of NPs in the pathophysiology of heart failure and particularly their counterbal­
ancing action opposing the overactivation of renin-angiotensin-aldosterone and sympathetic nervous systems has been described. In addition, NPs
are used today as key biomarkers in cardiovascular diseases with both diagnostic and prognostic significance. On these premises, multiple therapeutic
strategies based on the biological properties of NPs have been attempted to develop new cardiovascular therapies. Apart from the introduction of
the class of angiotensin receptor/neprilysin inhibitors in the current management of heart failure, novel promising molecules, including M-atrial natri­

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


uretic peptide (a novel atrial NP-based compound), have been tested for the treatment of human hypertension. The development of new drugs is
currently underway, and we are probably only at the dawn of novel NPs-based therapeutic strategies. The present article also provides an updated
overview of the regulation of NPs synthesis and secretion by microRNAs and epigenetics as well as interactions of cardiac hormones with other
endocrine systems.
.............................................................................................................................................................................................
Keywords Cardiac natriuretic peptides • Secretion • Epigenetics • Cardiovascular diseases • Biomarkers • Therapy

with relevant consequences on BP, blood volume, and electrolyte


Historical landmarks of the homeostasis.10
endocrine heart After the discovery of ANF, subsequently re-named atrial natriuretic
Following the development of the concept of endocrine glands in the se­ peptide (ANP), other biologically active peptides of the natriuretic peptide
cond half of the XIX century, through the pioneering discoveries of (NP) family, the brain NP (BNP) and the C-type NP (CNP) were identified.
Claude Bernard on the glucose secretion by the liver into the portal ANP and BNP are synthetized mainly in the heart and to a lesser extent in
vein ‘milieu interieur’1 and of the studies of Thomas Addison in adrena­ other organs.16 CNP is mainly produced by endothelial cells (ECs) and
lectomized animals,1 in 1898 Camillo Golgi described cytoplasmic bod­ lacks the carboxyl-terminal of ANP and BNP.17–19 Although BNP was ori­
ies containing hormones.2 In the early ’50s, Jameson and Palade ginally isolated from porcine brain, is mostly expressed in the cardiac atria.
established the function of the Golgi apparatus in secretory protein traf­ ANP and BNP produce similar endocrine responses promoting intense
ficking,3 while Kisch4 in 1956 demonstrated the presence of the secre­ natriuretic/diuretic effects, while CNP is mostly involved in regulating
tory granules in mammalian atria. At the same time, Gauer and Henry blood vessel tone through a paracrine function.19
demonstrated that the cardiac atria could sense the ‘fullness’ of the car­
diovascular system and produce adjustments in renal salt and water ex­
cretion to maintain a constant blood volume.5 In their experiments, The heart as an endocrine organ
stretching the atrium by balloon inflation of anaesthetized dogs resulted
Beside the production and secretion of NPs, multiple substances acting as
in enhanced diuresis and natriuresis in animals kept on parallel
endocrine and paracrine hormones are released by cardiomyocytes and
circulation.5
other cell types within the heart.20–22 Growth differentiation factor 15
Based on this integrated evidence, the ‘smoking gun’ of the heart
(GDF-15), a divergent member of the transforming growth factor
functioning as an endocrine gland was under the eyes. The discovery
(TGF)-β family, once released into the circulation exerts, among other
of cardiac hormones had to wait three more decades, until in 1981
functions, the control of body growth through the glial derived neuro­
Adolfo De Bold and colleagues reported that atrial extracts contained
trophic factor receptor α–like (GFRAL). Myostatin, a member of the
an active peptide, initially named as atrial natriuretic factor (ANF),
TGF-β superfamily, acts as an endocrine hormone targeting skeletal muscle
which was able to promote rapid and massive diuresis and natriuresis to modulate muscle mass through activin receptors. Both hormones, de­
when injected in rats.6–8 Several studies demonstrated that the admin­ riving from pre-propeptides, are increased in coronary heart disease
istration of ANF produced significant haemodynamic effects, such as (CHD) and HF.21 Other factors released from the heart, including pro­
vasodilatation, blood pressure (BP) reduction, haematocrit increase teins, lipids, and small molecules such as microRNAs (miRNAs) and exo­
and increased glomerular filtration rate, in both animals and hu­ somes, mainly exert autocrine/paracrine functions.21 In fact, they
mans.9–12 ANF antagonized angiotensin II-induced contraction in iso­ contribute to regulate myocyte growth, inhibition of fibroblast prolifer­
lated aorta, explaining the greater sensitivity to the BP-lowering ation and extracellular matrix deposition, coronary endothelium, and vas­
effect of ANF in renin-dependent hypertension with elevated levels cular smooth muscle cell (VSMC) proliferation and contractility,
of angiotensin-II and aldosterone.9,13 In humans, ANF lowered aortic myocardial perfusion, and vascular remodelling. In the specific, GDF-15
pressure in essential hypertension.14 In heart failure (HF) patients, and follistatin (both members of the TGF-β family) reduce the ischae­
ANF infusion increased absolute and fractional sodium excretion and mia/reperfusion injury and the pressure overload-induced hypertrophy.
urine flow rate and reduced plasma renin and aldosterone levels.15 The fibroblast growth factors (FGFs) decrease cardiac fatty acid oxidation
As a result of these pioneering studies, the main endocrine cardiovas­ and inhibit excessive autophagy. Endothelin-1 (ET-1) and activin-A pro­
cular functions of these cardiac hormones were progressively charac­ mote cardiomyocyte survival. The soluble suppression of tumourigenesis-2
terized and identified in increased natriuresis, diuresis and vasorelaxation and FGFs prevents cardiac fibrosis.21,23–28
Natriuretic peptides in clinical practice 645

The regulation of gene expression, left atrium along with an increase in circulating BNP, but they remained
below or at the limit of detection in the LV. In overt HF, BNP mRNA
secretion, and clearance of was further increased in the left atrium and in the LV, together with
natriuretic peptides a BNP increase in LV and blood, probably due to an additional recruit­
ment of ventricular BNP expression.40 These data suggest that the atrial
The hallmarks of ANP and BNP production by cardiomyocytes include secretion of BNP might not be sufficient and adequate to counteract
regulated secretion from the secretory granules and proteolytic pro­ pressure and volume increases as well as hyperactivation of sodium-
cessing leading from precursors to mature and biologically active pep­ retaining and vasopressor neurohormones during HF progression.
tides or inactive fragments17 (Figure 1). In patients who had undergone orthotopic heart transplantation
Cardiomyocytes store prohormones, namely proANP and proBNP, (with the native atria being left), plasma levels of NPs were not normal­
in secretory granules. These pro-peptide hormones are enzymatically ized by the replacement of the diseased ventricles even after the nor­
cleaved by proprotein convertases (corin and furin) and processed to malization of intracardiac pressures. This suggests that the atria are
form the N-terminal (NT) peptides NT-proANP and NT-proBNP the main source of NPs also in advanced HF41,42 and that the ventricular

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


and the biological active free carboxyl group (COOH)-terminal pep­ production is limited to advanced pathological conditions and strongly
tides ANP and BNP29,30 (Figure 1). Further processing of depends on ventricular alterations such as those observed in hyper­
NT-proANP generates the release of three biologically active frag­ trophy, fibrosis, inflammation, and hypoxia. In the latter context, the
ments, namely proANP1–28 (long-acting NP), proANP31–67, and hypoxia-inducible factor-1a, produced under low oxygen conditions,
proANP78–98.31,32 Differently from ANP, human proBNP shows post- activates transcription of both NPPA and NPPB, the genes encoding
translational glycosylation within the amino-terminal region.33 ANP and BNP, respectively.43
The binding of COOH-terminal peptides with type C NP receptor In addition to mechanical stimuli, several neurohormonal triggers may
(NPRC) is followed by internalization and lysosome degradation. The neu­ contribute to the secretion of NPs such as angiotensin II, adrenergic ago­
tral endopeptidase neprilysin (NEP) and the kidneys contribute to the NPs nists, ET-1, arginine vasopressin, glucocorticoids, thyroid hormones, sex
clearance. Due to its longer half-life compared to ANP (12–20 vs. 0.5– steroids, growth factors, and cytokines (tumour necrosis factor, interleu­
4 min), BNP exhibits enhanced natriuretic and diuretic actions. kins −1 and 6), further supporting the role of the heart as a component
In normal human plasma, the major circulating form of ANP is the of the endocrine system.44,45 More recently, an involvement of glucagon-
COOH-terminal peptide (αANP). The unprocessed proBNP circulates like peptide-1, one of the incretin hormones secreted by the epithelium
in plasma along with BNP and NT-proBNP. Several studies documen­ of the small intestine, has been implicated in the regulation of cardiac
ted that black individuals carry lower circulating levels of both ANP secretion.46
NT-proBNP and BNP and of midregional ANP (MRproANP), demon­ The abovementioned factors can regulate the expression of both
strating a state of NP deficiency in the Black race as a likely consequence NPPA and NPPB through the same transcriptional factors, including
of increased clearance rather than reduced synthesis.34,35 the rat sarcoma (RAS)/rapid accelerated fibrosarcoma (c-Raf-1) and
Many mechanisms contribute to the synthesis and secretion of ANP the phosphoinositide signalling pathways which stimulate the
and BNP. Acute (hours), subacute (days), and chronic (weeks) haemo­ p38-mitogen-activated protein kinase.47
dynamic and neurohormonal stimuli produce a different response.36 An emerging issue is represented by the epigenetic regulation of NPs
Acute mechanical stretches stimulate a phasic, short-term burst of synthesis. Different studies have detected epigenetic changes, consisting
NPs secretion from the existing atrial pool (regulated secretion), with­ in histone acetylation and methylation, in adult cardiomyocytes exposed
out clear effects on their synthesis.36 In subacute haemodynamic load to pathological conditions.48 Within the heart, both pressure and vol­
conditions, circulating NPs derive from both stored and newly synthe­ ume overloads have been associated with an increased acetylation of
sized hormones (regulated and constitutive secretion). In conditions of H3 and H4 histones and with demethylation of H3K9 at both NPPA
chronic haemodynamic overload, such as hypertension and HF, synthe­ and NPPB.49,50 The histone demethylase JMJD2A modulates NPPB in
sis of ANP and BNP occurs both in atria and ventricles.37 angiotensin II and ET-1 induced cardiac hypertrophy in human-induced
Regarding the cellular sites of NPs production, earlier data showed pluripotent stem cells.51 A reduced expression of three histone
that the ventricles do not normally contain large amounts of ANP or methylation-related molecules (H3K4me3, H3K9me2, and H3K9me3)
BNP, except some areas restricted to the proximal Purkinje system.6 was detected in non-ischaemic dilated cardiomyopathy.52
Subsequent studies showed that NPs synthesis and secretion may be DNA methylation may affect gene function and repress transcription by
differently regulated in atrial compared to ventricular cardiomyocytes altering promoter DNA accessibility and blocking the binding of transcrip­
and in neonatal vs. adult life. Indeed, electron microscopy studies de­ tion activating proteins.53–55 Aberrant DNA methylation of NPPA may par­
monstrated that secretory granules are not displayed in ventricular car­ ticipate in the mechanisms of cardiovascular disease (CVD), and it may
diomyocytes from normal hearts, whereas they may be observed in serve for the identification of individuals at high risk for CVD for primary
samples of ventricular myocardium collected during surgery or endo­ prevention and as a potential molecular target for therapeutic purposes.
cardial biopsies in patients with cardiac disease,38 thus suggesting a pre- Interesting data have been reported regarding the regulation of NPs by
programmed expression of foetal genes in ventricular cardiomyocytes microRNAs.56 Three miRNAs are known to modulate NPPA expression:
under pathological conditions. Cardiac catheterization data in humans miR-425, miR-155, and miR-105.57–59 They act in the presence of the wild
with left ventricular (LV) hypertrophy showed that the abnormal type gene sequence but not when a single base-pair mutation falls within
NPs circulating levels were significantly derived from atrial sources.39 the miRNA binding site. Consequently, higher ANP levels are detected
Morphological data from immunocytochemistry and in-situ hybridiza­ in individuals carrying the minor allele at one of these sites such as the min­
tion studies in animal models supported the view that NPs production or G allele at rs5068, which does not bind miR-425.48 The higher ANP
is normally restricted to the atria and, more specifically, to the auricular plasma level detected in carriers of the minor G allele at rs5068 was related
appendages. In an experimental dog model of early LV dysfunction, to lower BP level, lower risk of hypertension, lower body mass index and
BNP mRNA and tissue BNP content were markedly increased in the waist circumference, lower prevalence of obesity, and metabolic
646 M. Volpe et al.

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


Figure 1 Secretion of atrial natriuretic peptide and brain natriuretic peptide in cardiomyocytes takes place through both regulated pathways from
mature secretory granules stored in the cytoplasm and the constitutive pathway involving a de novo peptide synthesis and the release of immature
granules into the cytoplasm. The N-terminal pro-natriuretic peptides processing through corin and furin allow the production of the amino terminal
peptides and the carboxyterminal peptides forms that circulate in the blood.

syndrome.60–62 An increased expression of cardiac miR-425 and of its cir­ ischaemic function and regulate myocardial perfusion and remodel­
culating level has been reported in Blacks as a likely contributor to their basal ling.65 A newly discovered autocrine property of ANP is the regulation
NP-deficient state and to the abnormal response to a high-carbohydrate of cardiac autophagy, a process devoted to the removal of damaged
challenge.35 In fact, the decrease of plasma MRproANP, detected in re­ cells and organelles, to protect the heart from ischaemic insult.66
sponse to a metabolic perturbation such as the high-carbohydrate chal­ These functions, similarly to the endocrine functions, are mediated
lenge, was significantly reduced in Black compared to White individuals.35 through the type A NP receptor (NPRA)67,68 (Figure 2).
NPPA AS1 belongs to the class of natural antisense transcripts and, The genetic ablation of either NPPA or Npr1 (the gene encoding
as a long noncoding RNA, overlaps the coding protein gene but is NPRA) produced cardiac hypertrophy in mice.69,70 Consistently, genet­
transcribed from the antisense strand. In fact, it inhibits the transcrip­ ic variants of NPPA responsible of reduced ANP expression were asso­
tion of the corresponding sense mRNA behaving as a natural feedback ciated to cardiac hypertrophy in humans.71 A marked cardiac fibrosis
mechanism. Consistently, NPPA-AS1 inhibition produced increased was detected in mice lacking NPPB.72 Conversely, high concentrations
plasma ANP level, increased renal cyclic guanosine-monophosphate of NPs modulate cardiomyocyte numbers during development oppos­
(cGMP) level, and reduced BP level in mice.63 This approach, that ing cardiomyocyte proliferation.73
has relevance during mammalian foetal heart development, in condi­ ANP also exerts anti-inflammatory, proliferative, pro-angiogenic, and
tions of biomechanical stress and in HF, may reveal useful for anti-apoptotic effects within the endothelium.74 Lack of ANP as well as
therapeutic purposes. An additional mechanism of NPPA modulation a structurally altered ANP variant derived from a human gene mutation
depends on a corin-miRNA-1–3p axis responsible of a potent inhib­ associate with impaired EC viability and proliferation.75 BNP, produced
ition of corin activity and therefore of NPPA transcription, as shown by satellite cells within ischaemic skeletal muscle or by cardiomyocytes
by Celik et al.64 in response to pressure load, promotes endothelial protection via
NPRA.76 More recently, the modulation of pericytes and microcircula­
tory tone within the kidneys has been associated to the acute and
Autocrine/paracrine functions of chronic modulation of arterial BP by ANP.77
natriuretic peptides within the CNP plays a physiological autocrine role in cardiovascular homeosta­
sis by acting at the vascular level through the type B NP receptor
heart and vasculature (NPRB).78 It is also known as a key hormone for bone growth, as shown
Beside the natriuretic, diuretic and vasorelaxant effects, NPs exert by the systemic CNP KO mice.79 CNP is a major member of the
autocrine/paracrine functions. In the heart, they contribute to the regu­ non-NO, non-prostanoid component of endothelium-dependent re­
lation of myocyte growth, inhibition of fibroblast proliferation and laxation in the resistance vasculature. Endothelium-derived CNP con­
extracellular matrix deposition. They exert cytoprotective anti- tributes to the chronic regulation of vascular tone and systemic BP
Natriuretic peptides in clinical practice 647

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


Figure 2 The signalling pathways of the three natriuretic peptide receptors are represented. Type A and B natriuretic peptide receptors stimulate the
formation of cyclic guanylate monophosphate through the activation of particulate guanylate cyclase. The stimulation of guanylate cyclase activity takes
place also through the NO-soluble guanylate cyclase pathway. The two pathways do not necessarily share common molecular mechanisms, though the
final effects coincide. Cyclic guanylate monophosphate produces PKG, CNG, and PDEs. While PKG allows the biological functions of natriuretic pep­
tides in target tissues, phosphodiesterases metabolize cyclic guanylate monophosphate and reduce its effects. Activation of type C natriuretic peptide
receptor by C-type natriuretic peptide leads to the inhibition of adenylate cyclase, stimulation of phospholipase C, extracellular signal regulated kinase,
and phosphoinositide 3-kinase/protein kinase B. These pathways mediate the cardiac and vascular effects of C-type natriuretic peptide described by
references79–83. The final cardiovascular and renal effects of natriuretic peptides mediated by the three receptors are reported in the figure.
Abbreviation: CNG, gated-ion channel; PKG, protein kinase G; NO, nitric oxide.

by maintaining endothelial function and integrity independently of A still unexplained issue is represented by the lower NPs circulat­
NPRB.80 Moyes et al.81 underscored the relevance of NPRC signalling ing levels found in obese compared with lean healthy subjects. An in­
as the main mechanism underpinning CNP-dependent vascular pro­ creased expression of the NPRC in fat tissue can contribute to
tective functions (Figure 2). Consistently, the administration of NPRC explain this finding. The increased production of some adipokines re­
agonists promoted vasorelaxation in isolated resistance arteries and a leased by visceral fat tissue may also play a role. Finally, reduced car­
BP reduction that was not observed in mice lacking NPRC. Within diac secretion of NPs or impaired synthesis may contribute to lower
the heart, CNP derived from ECs, cardiomyocytes, and fibroblasts co- plasma NP levels in obesity.87 Moreover, ANP potentiates the effect
ordinates and preserves cardiac structure, function, and coronary va­ of glucagon-like peptide 1 (GLP-1) on glucose-induced insulin
soreactivity via activation of NPRC.82 secretion.88
Regarding the interaction between the NP system and steroid hor­
mones, experimental studies demonstrated that female sex steroids in­
Interactions between cardiac and crease NPPA expression in a dose-dependent manner and that both
oestradiol and progesterone are necessary to maintain suitable levels
other endocrine systems of NPPA expression in rat cardiomyocytes. On the other hand, the
An interaction between the NPs and adipose tissue has been consistent­ ANP concentration and atrial stores were increased in castrated
ly described. Indeed, the antihypertrophic effect of some adipokines, male rats. The testosterone replacement was associated with a de­
such as leptin, resistin, and visfatin, is linked to a stimulatory action on crease of plasma ANP concentration, but not of atrial stores. The ex­
ANP and BNP secretion in the heart of ob/ob mice.83 In turn, NPs perimental data suggest that both female and male sex steroid
play a role in the regulation of fat tissue function and growth by activating hormones influence NPs secretion, although with opposing effects.89
lipolysis, enhancing lipid oxidation, postprandial energy expenditure, and Moreover, hormone replacement therapy has shown a stimulatory ac­
mitochondrial respiration.84 NPs also exert metabolic actions and acti­ tion on the secretion of NPs in postmenopausal women.90 Regarding
vate the co-transcriptional activator peroxisome proliferator-activated- adrenal corticosteroids, an inverse relationship with NPs has emerged
receptor-G-coactivator (PGC)-1α and the proliferator-activated from different studies. While corticosteroids have direct and indirect
receptor-d, promoting muscle mitochondrial biogenesis and fat stimulating actions on cardiac endocrine function, NPs inhibit the secre­
oxidation.85 Recently, CNP has emerged as a regulator of energy metab­ tion of corticosteroids both directly, through the specific receptors on
olism through the control of adipogenesis, thermogenesis, and glucose zona fasciculata and glomerulosa cells, and indirectly by inhibiting the
clearance through NPRB and NPRC, triggering increases in cGMP and RAS.91 ANP is more potent than BNP in inhibiting the aldosterone re­
decreases in cAMP, respectively.86 sponse to angiotensin-II.92
648 M. Volpe et al.

a miR30-GALNT1/2 axis is a mechanism responsible of the increased


secretion of inactive proBNP in HF, likely contributing to the NPs re­
sistance and to the disease progression.95
Regarding the mechanisms of resistance at the receptor level, an in­
creased expression of the NPRC in some peripheral tissues may play a
role, by reducing the circulating levels of active NPs. Other studies showed
that gene variants of NPs receptors may contribute to the development of
CVDs, such as cardiomyopathies and stroke.61,62 Downstream to the NP
receptors, cyclic nucleotide-dependent intracellular molecular pathways
are regulated by phosphodiesterases (PDEs), which metabolize the
cGMP (Figure 3). PDEs are involved in complex cellular responses and their
dysregulation may contribute to an increased resistance to NPs in CVDs.96
Based on both experimental and human studies, PDEs 1–5, 8, and 9 are

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


dysregulated in HF.36 The post-receptor regulation of NPs activity oper­
ates through the binding with the G-coupled receptors NPRA and
NPRB and the subsequent stimulation of a membrane-bound particulate
guanylate cyclase (pGC), thus improving endothelial function, reducing pul­
monary pressures and increasing cardiac output.97,98 In such a context,
Figure 3 Schematic representation of the interactions between
strategies based on activation/stimulation of the NO-soluble GC pathway
natriuretic peptides and phosphodiesterases. Cyclic guanosine-
might have synergistic effects with NPs in HF.
monophosphate stimulates the production of phosphodiesterase 2
and phosphodiesterase 5, whereas it antagonizes phosphodiesterase 3.
Also, phosphodiesterase 1 and phosphodiesterase 9 metabolize cyclic
guanosine-monophosphate. Cyclic adenylate monophosphate is mainly
Natriuretic peptides as markers
metabolized by phosphodiesterases 2, 3, 4, 8. of cardiovascular diseases
NPs have been validated as useful diagnostic and prognostic biomarkers
in CVDs. In addition to being well-established diagnostic and prognostic
Impaired endocrine response of the markers of HF, plasma levels of NPs have been proposed as biomarkers
also in atrial fibrillation, CHD, and valvular heart disease.
heart In the non-acute HF setting, the upper limits of normal levels are
The ‘endocrine paradox’ in HF is related to the observation that fluid 35 pg/mL for BNP, 125 pg/mL for NT-proBNP, and 120 pg/mL for
retention and vasoconstriction occur despite high or very high circulat­ mid regional-proANP (MR-proANP).99 BNP and NT-proBNP levels
ing NPs levels. Early evidence in the ’90s showed that plasma ANP levels are currently used in clinical practice to confirm the diagnosis of HF
were not properly increased by volume expansion with saline solution and are associated with disease severity. Based on knowledge that high­
in patients with dilated cardiomyopathy and mild cardiac systolic dys­ er NT-proBNP levels, besides reflecting more severe heart disease, de­
function, before the onset of signs or symptoms of overt HF. The in­ pend on age, female sex, and renal function, the range of normal levels in
appropriate increase of ANP levels might contribute to the abnormal older people and other clinical settings needs to be further defined.
vasoconstriction and water/sodium retention in HF patients exposed Table 1 summarizes available evidence regarding the diagnostic and
to acute or chronic volume overload.91 Moreover, failure to suppress prognostic role of NPs in CVDs.99–118
plasma renin activity in hypertensive patients in response to acute saline
load, along with an abnormal ANP response, was explained by an im­
paired relationship between increases of blood volume and those of at­ Role of natriuretic peptide–based
rial pressure or wall stress during saline infusion.93 Resistance to the
biological effects of NPs can be attributed to different mechanisms, act­
strategies to prevent heart failure
ing at pre-receptor, receptor, and post-receptor levels. Regarding the The use of BNP has been successfully introduced in programmes of HF
pre-receptor level, an impaired post-translational processing of prevention.119–121 The St. Vincent’s Screening to Prevent HF
proNPs has been hypothesized.48 Indeed, increased levels of circulating (STOP-HF) included asymptomatic individuals with one or more risk
non-biologically active proBNP-derived fragments have been identified factors of CVD. The trial compared a BNP-based screening based on
by chromatographic procedures in human plasma, consisting in the in­ a collaboration between primary care physicians and cardiologists.
tact and glycosylated forms of the precursor proBNP and The control group received the primary care physician and specialist
NT-proBNP.94 Furthermore, plasma corin levels are significantly lower care when required. The intervention group showed a reduction in
in HF patients than in controls with a relationship between reduced HF development.122 Consistently, a genetic sub-study of the
plasma enzyme levels and disease severity. Corin and furin cleavage STOP-HF trial revealed that the minor C allele of a BNP genetic variant,
of proBNP into active BNP could be suppressed by the glycosylation rs198389, associated with higher circulating BNP levels, was associated
of proBNP near to the cleavage site of the enzymes, which makes with lower risk of hypertension, new onset LV dysfunction or major ad­
the molecule resistant to proteolysis.30 In HF patients carrying a higher verse cardiovascular events.123 The PONTIAC study used the
proportion of proBNP a higher glycosylation rate at threonines 48 and NP-based approach as part of a strategy to identify subjects at the high­
71 with a consequent reduction of its processing was described. The est risk of cardiovascular events, targeting treatment to these groups to
GalNac-transferase (GALNT) 1 and 2 mediate the glycosylation- prevent HF and other CVDs. The study included 300 diabetic patients
regulated increase of proBNP secretion in these subjects. Of interest, free from CVD but with increased NT-proBNP level, who were
Natriuretic peptides in clinical practice 649

Table 1 Diagnostic and prognostic role of NPs in CVDs

Natriuretic peptide Clinical conditions


......................................................................................................................................................................................
NT-proBNP Diagnostic role in acute, subacute, and chronic HF99,100
ACS,101 pulmonary embolism,102 pulmonary hypertension103,104
Prognostic role for increased risk of stroke, systemic embolism, and cardiac-related death in patients with AF, improving the
CHA2DS2-VASc score105
Markers of cognitive decline and dementia consequent to brain microcirculatory damage and dysfunction in hypertensive
patients106
NT-proANP and Markers of progression of coronary atherosclerosis from moderate to severe101
NT-proBNP

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


NT-proANP and BNP Predictor of future CV events, including death and recurrent MI, in stable CAD patients101,107
Predictor of future CV events following coronary revascularization (PCI/stent)108,109
BNP Increased levels are associated with severity of aortic stenosis, LV chamber size, wall thickness and stress, LVEF, left atrial size,
and right ventricular pressure110
J-shaped relationship with all-cause and cardiovascular mortality rates in HFpEF patients undergoing TAVR111
Increased levels are directly related with mean pulmonary arterial pressure in patients with pulmonary hypertension112
NT-proBNP and BNP Increased levels assess the progression of mitral valve disease, contribute to more accurate risk stratification by identifying
patients who are more likely to experience death from CV causes, HF, and cardiac hospitalizations113
NT-proANP Prognostic role towards digital ulcers development and mortality in systemic sclerosis patients114,115
MR-proANP Diagnostic role in acute and sub-acute HF116 Prognostic role in HCM117
MR-proANP combined with Improved prognostic role in HF118
BNP

ACS, acute coronary syndrome; ANP, atrial natriuretic peptide; BNP, brain natriuretic peptide; CAD, coronary artery disease; CV, cardiovascular; HCM, hypertrophic cardiomyopathy; HF, heart
failure; HFpEF, heart failure with preserved ejection fraction; LV, left ventricle; LVEF, left ventricular ejection fraction; MR-proANP, mid-regional pro-atrial natriuretic peptide; NT-proANP,
N-terminal pro-atrial natriuretic peptide; NT-proBNP, N-terminal pro-brain natriuretic peptide; PCI, percutaneous coronary intervention; TAVR, transcatheter aortic valve replacement.

randomized to either standard treatment or an intensified strategy in­ NPs activity.127 After a series of unsuccessful attempts by using NPs ana­
volving a cardiac outpatient clinic. As a result, the intensified treatment logues with longer half-life of the naturally occurring NPs, NEP inhibitors
led to a significant reduction of CVD development, HF, and hospitaliza­ alone, or in combination with ACE-inhibitors, the pharmacological regu­
tions.124 Interestingly, the incorporation of NPs levels to clinical infor­ lation of the endocrine function of the heart has resulted in the thera­
mation led to superior risk prediction of HF, compared to a model only peutic combination of the angiotensin receptor inhibitor valsartan
incorporating risk factors, among individuals with dysglycaemia.124 with the NEP inhibitor (ARNi) sacubitril. NEP is also involved in the deg­
Both the STOP-HF and PONTIAC trials are included in the 2014 radation of other biological active peptides including bradykinin and
Canadian Cardiovascular Society HF Management Guidelines to sup­ angiotensin II.128 Therefore, the concomitant inhibition of type 1 angio­
port a recommendation for the use of NPs in at-risk individuals.125 tensin II receptor (AT1R) and of NEP, achieved by combining sacubitril
These guidelines recommend that NPs levels should be used to imple­ and valsartan, produces a synergistic effect by increasing NPs levels and
ment strategies to prevent HF in individuals with risk factors. reducing the angiotensin II-related signal transduction pathways.129
Based on the high diagnostic and prognostic value of NPs levels in the Experimental studies assessed the impact of ARNi on the circulating
preclinical stage of HF and other CVDs, the use of NPs measurement in
NPs levels.130 A human study by Murphy et al.131 considered 23 patients
the clinical practice should be recommended.
with mild to moderate HF with reduced ejection fraction (HFrEF) and
Beside their role as biomarkers of HF severity, the results of different
measured BNP using 5 different assays, NT-proBNP using 3 assays,
studies have suggested that serial measurements of NPs may be helpful
and ANP, MR-proANP, proBNP,1–107,132 and CNP using a single assay
to guide titration of long-term medical therapy in HF. Indeed, it has
before and 22, 46, and 84 days after initiating sacubitril/valsartan therapy.
been shown that patients randomized to receive biomarker-guided
As a result, sacubitril/valsartan led to small and inconsistent changes in
therapy achieved a greater decrease in NPs levels over time compared
with those kept in the usual care strategy, these changes being asso­ BNP (that varied across the methods), to a decrease of variable extent
ciated with improvements in clinical outcomes.126 of MR-proANP, NT-proBNP, and proBNP,1–107,132 whereas CNP levels
did not show any consistent change. The most striking result, consistent
Current therapeutic implications of with the experimental evidence, was a highly significant increase of
αANP, suggesting a role for this peptide. N-terminal peptides are char­
cardiac endocrine function acterized by major stability and, since they are not cleared by NEP, are
For many years, the potential advantages of NPs properties in HF have the only measurable fragments during ARNi treatment. Since the publi­
encouraged the development of drugs which could mimic or enhance cation of the proof-of-concept study PARADIGM-HF (Angiotensin
650 M. Volpe et al.

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


Figure 4 The impact of neprilysin on atrial natriuretic peptide, brain natriuretic peptide, and C-type natriuretic peptide metabolism ultimately in­
creases the level of the carboxyterminal peptides in the circulation with consequent beneficial effects for the cardiovascular system. The decrease
of the circulating amino terminal peptides observed upon ARNi administration mirrors the improvement of cardiac function. The increased brain natri­
uretic peptide level, such as that occurring in chronic heart failure, has been suggested to behave as an endogenous inhibitor of neprilysin.

Receptor-Neprilysin Inhibitor with ACEI to Determine Impact on Global recent post-hoc analysis of the study including only investigator-
Mortality and Morbidity in HF),133 the benefits of sacubitril/valsartan in identified events.145
chronic and acute HFrEF setting have been consistently demon­
strated.134–137 More recently, sacubitril/valsartan has been associated
with a significant reduction in CV mortality and HF hospitalizations across Future perspectives for therapeutic
a wide range of the ejection fraction (EF) spectrum.138,139 implications of natriuretic peptides
Based on this evidence, European Guidelines recommend sacubitril/val­
sartan as first-line treatment in HFrEF and suggest considering its use in HF
in cardiovascular diseases
with mildly reduced EF (40%–49%).99 Moreover, US Food and Drug Experimental studies in mice showed that the NPPA knockout led to salt-
Administration approved an expanded indication for sacubitril/valsartan sensitive hypertension and lack of Npr1 associated with salt-sensitive
in HF with preserved EF (HFpEF) patients or independently of LVEF.140 hypertension. Intravenous ANP administration in a model of
In contrast with the above-mentioned studies, patients with chronic EC-specific, but not VSMC-specific, deletion of Npr1 resulted in dimin­
advanced HFrEF included in the LCZ696 (sacubitril/valsartan) in the ished BP reduction, highlighting the role of endothelial NPRA for BP and
Hospitalized Advanced Heart Failure (LIFE) trial did not significantly re­ vascular tone regulation by ANP.146 In humans, molecular genetic stud­
duce the NT-proBNP level when treated with sacubitril/valsartan com­ ies have revealed that reduced NPs expression contributes to an in­
pared to valsartan.141 Among other plausible explanations, a creased occurrence of hypertension, these results being confirmed by
BNP-mediated NEP inhibition might have contributed to the unexpect­ genome-wide association studies23 and by the evidence that primary
ed results. Indeed, based on a previous study, BNP levels >916 pg/mL hypertension is a condition of ANP deficiency.147 Based on this patho­
behave as an endogenous NEPi.142 Thus, NEP might be already inhibited physiological and genetic background, several NP-based therapeutic ap­
by high BNP level in patients with advanced HF, without any additional proaches, including ANP-recombinant peptides (carperitide and
benefits upon sacubitril/valsartan treatment143 (Figure 4). If this hypoth­ ularitide), endothelin converting enzyme/neprilysin inhibitors, and
esis is corroborated by more direct evidence, it may support alternative ARNi have been developed in hypertension.148 However, no significant
strategies to enhance the NP actions, such as the stimulation of NP benefits were obtained, and an excess of adverse events was reported.
receptors. More recently, a novel ANP-based compound (M-atrial natriuretic pep­
In the PARADISE-MI (Prospective ARNI vs. ACE Inhibitor Trial to tide [MANP]), a potent NPRA stimulator with a greater resistance to
Determine Superiority in Reducing Heart Failure Events after enzymatic degradation by both NEP and insulin degrading enzyme and
Myocardial Infarction) study conducted in patients with acute myocar­ reduced removal by NPRC, has shown long lasting and dose-dependent
dial infarction no significant differences in the composite outcome of BP lowering effect and no relevant short-term adverse effects.149,150
cardiovascular death, HF hospitalizations or outpatient development MANP, a 40 amino acid peptide compared to 28 amino acids ANP, car­
of HF were found between sacubitril/valsartan and ramipril.144 ries a natural peptide mutation firstly discovered in a family affected by
However, a superiority of sacubitril/valsartan has been reported in a AF.151 The clinical development of this innovative NP-based approach
Natriuretic peptides in clinical practice 651

Table 2 summarizes the main therapeutic strategies based on inter­


Table 2 Therapeutic strategies based on NPs
ventions on the NP system.48–52,57,63,98,133–139,149–151,155–169
Level of Therapeutic strategies
intervention
.................................................................................... Conclusions
NPPA and NPPB NPPA-AS1 (long noncoding RNA and natural
expression antisense transcript for NPPA)63 Studies performed in the last 40 years have consistently supported that
the heart has an endocrine function exerted through the action of dif­
microRNAs inhibiting the NPPA expression:
ferent hormones. The NP system appears to play a prominent role in
miR-425, miR-155, miR-10557–59
physiological and pathological cardiovascular conditions and to interact
Histone acetylation and DNA methylation48–52 with extracardiac hormonal systems.
Reduction of NEPi (Candoxatril)155 The three main components of the NP system act through specific
NPs receptors and intracellular signalling pathways. ANP and BNP are most­
ACE/NEPi (Omapatrilat)156

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


degradation ly involved in the control of cardiovascular homeostasis through their
ARB/NEPi (Sacubitril/valsartan)133–139 natriuretic, diuretic, and vasorelaxant effects. CNP is mostly involved
ECE/NEPi (Daglutril)157 in regulating vascular tone.
NPR activation Nesiritide (synthetic human BNP)158
Due to their biological properties, components of the NP systems
have been largely validated as useful diagnostic and prognostic biomar­
Carperitide, Ularitide (recombinant human ANP)159 kers in different CVDs. NP-based therapeutic approaches might re­
Cenderitide (CD-NP)160 present promising future strategies contributing to enhance the
Vosoritide (analogue of human CNP)161 beneficial effects of an efficacious neurohormonal inhibition and to
counteract the development and progression of HF and other CVDs.
Designer peptides: MANP149–151 ASBNP.1 (derived
from an alternative spliced transcript form of
BNP)162 Conjugated ANP Funding
(ANP-Fc, ANP-HSA, CNP/ANP)163,164 All authors declare no funding for this contribution.
Triazine derivatives (GC-A non-peptide Conflict of interest: M.V. reports personal fees for speaker bureau and/
activators)165 or consulting in Advisory Board from Amarin, Amgen, Astra Zeneca, Kalos
Activating antibodies163 Medical, Menarini Int, Novartis Pharma, Novo Nordisk, outside the submit­
ted work. S.R. and G.G. have nothing to disclose.
GC-A positive allosteric modulators
(MCUF-651)166
Post-receptor PDE inhibitors167
Data availability
enhancers
There are no new data associated with this article.
sGC stimulators (vericiguat)98,168
sGC activators169
References
ACE, angiotensin converting enzyme; ANP, atrial natriuretic peptide; ANP-Fc, ANP 1. Eknoyan G. Emergence of the concept of endocrine function and endocrinology. Adv
fused to the Fc domain of IgG; ANP-HSA, recombinant human ANP and human Chronic Kidney Dis 2004;11:371–376. https://doi.org/10.1053/j.ackd.2004.07.003
serum albumin; ARB, angiotensin receptor blocker; BNP, brain natriuretic peptide; 2. Dröscher A. Camillo Golgi and the discovery of the Golgi apparatus. Histochem Cell
ASBNP.1, alternatively spliced; CNP, C-type natriuretic peptide; ECE; Biol 1998;109:425–430. https://doi.org/10.1007/s004180050245
endothelin-converting enzyme; GC-A, guanylyl cyclase A receptor; MANP, novel 3. Weibel ER, Palade GE. New cytoplasmic components in arterial endothelia. J Cell Biol
ANP-based compound; miR; microRNA; NEPi; neprilysin inhibitor; NPs, natriuretic 1964;23:101–112. https://doi.org/10.1083/jcb.23.1.101
peptides; NPPA, gene encoding ANP; NPPB, gene encoding BNP; NPR, natriuretic 4. Kisch B. Electron microscopy of the atrium of the heart. I. Guinea pig. Exp Med Surg
peptide receptor; PDE, phosphodiesterase; sGC, soluble guanylate cyclase. 1956;14:99–112.
5. Henry JP, Gauer OH, Reeves JL. Evidence of the atrial location of receptors influencing
urine flow. Circ Res 1956;4:85–90. https://doi.org/10.1161/01.RES.4.1.85
6. de Bold AJ, Borenstein HB, Veress AT, Sonnenberg H. A rapid and potent natriuretic
response to intravenous injection of atrial myocardial extract in rats. Life Sci 1981;28:
may find a role in the management of hypertension and may contribute 89–94. https://doi.org/10.1016/0024-3205(81)90370-2
to prevent the development of organ damage, HFrEF and HFpEF.152 7. De Bold AJ, Bencosme SA. Studies on the relationship between the catecholamine dis­
tribution in the atrium and the specific granules present in atrial muscle
Another fragment derived from the NT-proANP, the proANP 31–67
cells. 1. Isolation of a purified specific granule subfraction. Cardiovasc Res 1973;7:
peptide, has shown cardiorenal protective actions, independently 351–363. https://doi.org/10.1093/cvr/7.3.351
from the NPR/cGMP pathway, in a preclinical model of maladaptive car­ 8. De Bold AJ. Heart atria granularity effects of changes in water-electrolyte balance. Proc
diac hypertrophy and renal damage induced by hypertension.153 It ap­ Soc Exp Biol Med 1979;161:508–511. https://doi.org/10.3181/00379727-161-40584
9. Volpe M, Atlas SA, Sosa RE, Marion DE, Mueller FB, Sealey JE, et al. Angiotensin
pears a promising therapeutic tool to counteract hypertensive organ II-induced atrial natriuretic factor release in dogs is not related to hemodynamic re­
damage, renal diseases, and HF.32 sponses. Circ Res 1990;67:774–779. https://doi.org/10.1161/01.RES.67.3.774
Thanks to the growing knowledge on synthesis, secretion, and regu­ 10. Volpe M, Vecchione F, Cuocolo A, Lembo G, Pignalosa S, Condorelli M, et al.
lation of NPs, new strategies can be developed. The genetic/epigenetic Hemodynamic responses to atrial natriuretic factor in nephrectomized rabbits: at­
tenuation of the circulatory consequences of acute volume expansion. Circ Res
modulation of NPs represents an attractive approach.154 Therefore, 1988;63:322–329. https://doi.org/10.1161/01.RES.63.2.322
potential RNA-based targets to achieve an increased level of circulating 11. Kleinert HD, Volpe M, Odell G, Marion D, Atlas SA, Camargo MJ, et al. Cardiovascular
NPs include miR-425 and miR-155.57 Furthermore, modulation of effects of atrial natriuretic factor in anesthetized and conscious dogs. Hypertension
1986;8:312–316. https://doi.org/10.1161/01.HYP.8.4.312
NPPA methylation may serve as a useful tool to change circulating
12. Volpe M, Odell G, Kleinert HD, Müller F, Camargo MJ, Laragh JH, et al. Effect of atrial
ANP levels. Currently, work is ongoing to elucidate the therapeutic natriuretic factor on blood pressure, renin, and aldosterone in Goldblatt hypertension.
benefit of NPPA-AS1 knock-down in models of HFpEF and HFrEF.63 Hypertension 1985;7:I43–I48. https://doi.org/10.1161/01.HYP.7.3_Pt_2.I43
652 M. Volpe et al.

13. Burnett JC Jr, Granger JP, Opgenorth TJ. Effects of synthetic atrial natriuretic factor on 38. Frustaci A, Russo MA, Chimenti C. Diagnostic contribution of left ventricular endo­
renal function and renin release. Am J Physiol 1984;247:F863–F866. https://doi.org/10. myocardial biopsy in patients with clinical phenotype of hypertrophic cardiomyopathy.
1152/ajprenal.1984.247.5.F863 Hum Pathol 2013;44:133–141. https://doi.org/10.1016/j.humpath.2012.04.023
14. Volpe M, Mele AF, Indolfi C, De Luca N, Lembo G, Focaccio A, et al. Hemodynamic and 39. Murakami Y, Shimada T, Inoue S, Shimizu H, Ohta Y, Katoh H, et al. New insights into
hormonal effects of atrial natriuretic factor in patients with essential hypertension. J the mechanism of the elevation of plasma brain natriuretic polypeptide levels in pa­
Am Coll Cardiol 1987;10:787–793. https://doi.org/10.1016/S0735-1097(87)80271-1 tients with left ventricular hypertrophy. Can J Cardiol 2002;18:1294–1300.
15. Cody RJ, Atlas SA, Laragh JH, Kubo SH, Covit AB, Ryman KS, et al. Atrial natriuretic 40. Luchner A, Stevens TL, Borgeson DD, Redfield M, Wei CM, Porter JG, et al.
factor in Normal subjects and heart failure patients. Plasma levels and renal, hormonal, Differential atrial and ventricular expression of myocardial BNP during evolution of
and hemodynamic responses to peptide infusion. J Clin Invest 1986;78:1362–1374. heart failure. Am J Physiol 1998;274:H1684–H1689. https://doi.org/10.1152/ajpheart.
https://doi.org/10.1172/JCI112723 1998.274.5.H1684
16. Volpe M, Rubattu S, Burnett J Jr. Natriuretic peptides in cardiovascular diseases: cur­ 41. Ogawa T, Veinot JP, Davies RA, Haddad H, Smith SJ, Masters RG, et al.
rent use and perspectives. Eur Heart J 2014;35:419–425. https://doi.org/10.1093/ Neuroendocrine profiling of humans receiving cardiac allografts. J Heart Lung
eurheartj/eht466 Transplant 2005;24:1046–1054. https://doi.org/10.1016/j.healun.2004.06.023
17. Levin ER, Gardner DG, Samson WK. Natriuretic peptides. N Engl J Med 1998;339: 42. Masters RG, Davies RA, Veinot JP, Hendry PJ, Smith SJ, de Bold AJ. Discoordinate
321–328. https://doi.org/10.1056/NEJM199807303390507 modulation of natriuretic peptides during acute cardiac allograft rejection in humans.
18. Lee CY, Burnett JC Jr. Natriuretic peptides and therapeutic applications. Heart Fail Rev Circulation 1999;100:287–291. https://doi.org/10.1161/01.CIR.100.3.287

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


2007;12:131–142. https://doi.org/10.1007/s10741-007-9016-3 43. Chun YS, Hyun JY, Kwak YG, Kim IS, Kim CH, Choi E, et al. Hypoxic activation of the
19. Goetze JP, Bruneau BG, Ramos HR, Ogawa T, de Bold MK, de Bold AJ. Cardiac natri­ atrial natriuretic peptide gene promoter through direct and indirect actions of
uretic peptides. Nat Rev Cardiol 2020;17:698–717. https://doi.org/10.1038/s41569- hypoxia-inducible factor-1. Biochem J 2003;370:149–157. https://doi.org/10.1042/
020-0381-0 bj20021087
20. de Bold AJ, Bruneau BG, Kuroski de Bold ML. Mechanical and neuroendocrine regula­ 44. Fish-Trotter H, Ferguson JF, Patel N, Arora P, Allen NB, Bachmann KN, et al.
tion of the endocrine heart. Cardiovas Res 1996;31:7–18. https://doi.org/10.1016/ Inflammation and circulating natriuretic peptide levels. Circ Heart Fail 2020;13:
S0008-6363(95)00121-2 e006570. https://doi.org/10.1161/CIRCHEARTFAILURE.119.006570
21. Zhao J, Pei L. Cardiac endocrinology: heart-derived hormones in physiology and dis­ 45. Richards AM. The renin-angiotensin-aldosterone system and the cardiac natriuretic
ease. JACC Basic Transl Sci 2020;5:949–960. https://doi.org/10.1016/j.jacbts.2020.05. peptides. Heart 1996;76:36–44. https://doi.org/10.1136/hrt.76.3_Suppl_3.36
007 46. Kim M, Platt MJ, Shibasaki T, Quaggin SE, Backx PH, Seino S, et al. GLP-1 receptor ac­
22. McGrath MF, de Bold ML, de Bold AJ. The endocrine function of the heart. Trends tivation and Epac2 link atrial natriuretic peptide secretion to control of blood pressure.
Endocrinol Metab 2005;16:469–477. https://doi.org/10.1016/j.tem.2005.10.007
Nat Med 2013;19:567–575. https://doi.org/10.1038/nm.3128
23. Eggers KM, Kempf T, Lagerqvist B, Lindahl B, Olofsson S, Jantzen F, et al.
47. Clerk A, Pham FH, Fuller SJ, Sahai E, Aktories K, Marais R, et al. Regulation of mitogen-
Growth-differentiation factor-15 for long-term risk prediction in patients stabilized
activated protein kinases in cardiac myocytes through the small G protein Rac1. Mol
after an episode of non-ST-segment-elevation acute coronary syndrome. Circ
Cell Biol 2001;21:1173–1184. https://doi.org/10.1128/MCB.21.4.1173-1184.2001
Cardiovasc Genet 2010;3:88–96. https://doi.org/10.1161/CIRCGENETICS.109.877456
48. Rubattu S, Stanzione R, Cotugno M, Bianchi F, Marchitti S, Forte M. Epigenetic control
24. Wollert KC, Kempf T, Wallentin L. Growth differentiation factor 15 as a biomarker in
of natriuretic peptides: implications for health and disease. Cell Mol Life Sci 2020;77:
cardiovascular disease. Clin Chem 2017;63:140–151. https://doi.org/10.1373/clinchem.
5121–5130. https://doi.org/10.1007/s00018-020-03573-0
2016.255174
49. Mahmoud SA, Poizat C. Epigenetics and chromatin remodeling in adult cardiomyop­
25. Ogura Y, Ouchi N, Ohashi K, Shibata R, Kataoka Y, Kambara T, et al. Therapeutic im­
athy. J Pathol 2013;231:147–157. https://doi.org/10.1002/path.4234
pact of follistatin-like 1 on myocardial ischemic injury in preclinical models. Circulation
50. Hohl M, Wagner M, Reil JC, Müller SA, Tauchnitz M, Zimmer AM, et al. HDAC4 con­
2012;126:1728–1738. https://doi.org/10.1161/CIRCULATIONAHA.112.115089
trols histone methylation in response to elevated cardiac load. J Clin Invest 2013;123:
26. Shen Y, Zhang X, Xu Y, Xiong Q, Lu Z, Ma X, et al. Serum FGF21 is associated with
1359–1370. https://doi.org/10.1172/JCI61084
future cardiovascular events in patients with coronary artery disease. Cardiology
51. Rosales W, Lizcano F. The histone demethylase JMJD2A modulates the induction of
2018;139:212–218. https://doi.org/10.1159/000486127
hypertrophy markers in iPSC-derived cardiomyocytes. Front Genet 2018;9:9–14.
27. Chou RH, Huang PH, Hsu CY, Chang CC, Leu HB, Huang CC, et al. Circulating fibro­
https://doi.org/10.3389/fgene.2018.00014
blast growth factor 21 is associated with diastolic dysfunction in heart failure patients
52. Ito E, Miyagawa S, Fukushima S, Yoshikawa Y, Saito S, Saito T, et al. Histone modifica­
with preserved ejection fraction. Sci Rep 2016;6:33953. https://doi.org/10.1038/
tion is correlated with reverse left ventricular remodeling in nonischemic dilated car­
srep33953
diomyopathy. Ann Thorac Surg 2017;104:1531–1539. https://doi.org/10.1016/j.
28. Planavila A, Redondo I, Hondares E, Vinciguerra M, Munts C, Iglesias R, et al. Fibroblast
growth factor 21 protects against cardiac hypertrophy in mice. Nat Commun 2013;4: athoracsur.2017.04.046
2019. https://doi.org/10.1038/ncomms3019 53. Marx A, Kahan T, Simon I. Integrative analysis of methylome and transcriptome reveals
29. Dietz JR. Mechanisms of atrial natriuretic peptide secretion from the atrium. Cardiovasc the importance of unmethylated CpGs in non-CpG island gene activation. Biomed Res
Res 2005;68:8–17. https://doi.org/10.1016/j.cardiores.2005.06.008 Int 2013;2013:785731. https://doi.org/10.1155/2013/785731
30. Semenov AG, Tamm NN, Seferian KR, Postnikov AB, Karpova NS, Serebryanaya DV, 54. Li J, Zhu J, Ren L, Ma S, Shen B, Yu J, et al. Association between NPPA promoter methy­
et al. Processing of pro-B-type natriuretic peptide: furin and corin as candidate conver­ lation and hypertension: results from Gusu cohort and replication in an independent
tases. Clin Chem 2010;56:1166–1176. https://doi.org/10.1373/clinchem.2010.143883 sample. Clin Epigenetics 2020;12:133. https://doi.org/10.1186/s13148-020-00927-0
31. Vesely DL. Discovery of new cardiovascular hormones for the treatment of congestive 55. Li J, Zhu J, Zhang Q, Chen L, Ma S, Lu Y, et al. NPPA promoter hypomethylation pre­
heart failure. Cardiovasc Hematol Disord Drug Targets 2007;7:47–62. https://doi.org/10. dicts central obesity development: a prospective longitudinal study in Chinese adults.
2174/187152907780059128 Obes Facts 2022;15:257–270. https://doi.org/10.1159/000521295
32. da Silva GJJ, Altara R, Booz GW, Cataliotti A. Atrial natriuretic peptide31–67: a novel 56. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;136:
therapeutic factor for cardiovascular diseases. Front Physiol 2021;12:691407. https:// 215–233. https://doi.org/10.1016/j.cell.2009.01.002
doi.org/10.3389/fphys.2021.691407 57. Arora S, Rana R, Chhabra A, Jaiswal A, Rani V. miRNA-transcription factor interac­
33. Matsuo A, Nagai-Okatani C, Nishigori M, Kangawa K, Minamino N. Natriuretic pep­ tions: a combinatorial regulation of gene expression. Mol Genet Genomics 2013;288:
tides in human heart: novel insight into their molecular forms, functions, and diagnostic 77–87. https://doi.org/10.1007/s00438-013-0734-z
use. Peptides 2019;111:3–17. https://doi.org/10.1016/j.peptides.2018.08.006 58. Arora P, Wu C, Hamid T, Arora G, Agha O, Allen K, et al. Acute metabolic influences
34. Gupta DK, Daniels LB, Cheng S, deFilippi CR, Criqui MH, Maisel AS, et al. Differences in on the natriuretic peptide system in humans. J Am Coll Cardiol 2016;67:804–812.
natriuretic peptide levels by race/ethnicity (from the multi-ethnic study of atheroscler­ https://doi.org/10.1016/j.jacc.2015.11.049
osis). Am J Cardiol 2017;120:1008–1015. https://doi.org/10.1016/j.amjcard.2017.06. 59. Wu C, Arora P, Agha O, Hurst LA, Allen K, Nathan DI, et al. Novel microRNA reg­
030 ulators of atrial natriuretic peptide production. Mol Cell Biol 2016;36:1977–1987.
35. Patel N, Russell GK, Musunuru K, Gutierrez OM, Halade G, Kain V, et al. Race, natri­ https://doi.org/10.1128/MCB.01114-15
uretic peptides, and high-carbohydrate challenge: a clinical trial. Circ Res 2019;125: 60. Newton-Cheh C, Larson MG, Vasan RS, Levy D, Bloch KD, Surti A, et al. Association of
957–968. https://doi.org/10.1161/CIRCRESAHA.119.315026 common variants in NPPA and NPPB with circulating natriuretic peptides and blood
36. Lugnier C, Meyer A, Charloux A, Andrès E, Gény B, Talha S. The endocrine function of pressure. Nat Genet 2009;41:348–353. https://doi.org/10.1038/ng.328
the heart: physiology and involvements of natriuretic peptides and cyclic nucleotide 61. Cannone V, Boerrigter G, Cataliotti A, Costello-Boerrigter LC, Olson TM, McKie PM,
phosphodiesterases in heart failure. J Clin Med 2019;8:174. https://doi.org/10.3390/ et al. A genetic variant of the atrial natriuretic peptide gene is associated with cardio­
jcm8101746 metabolic protection in the general community. J Am Coll Cardiol 2011;58:629–633.
37. Mangat H, de Bold AJ. Stretch-induced atrial natriuretic factor release utilizes a rapidly https://doi.org/10.1016/j.jacc.2011.05.011
depleting pool of newly synthesized hormone. Endocrinology 1993;133:1398–1403. 62. Cannone V, Cefalu’ AB, Noto D, Scott CG, Bailey KR, Cavera G, et al. The atrial natri­
https://doi.org/10.1210/endo.133.3.8365374 uretic peptide genetic variant rs5068 is associated with a favorable cardiometabolic
Natriuretic peptides in clinical practice 653

phenotype in a Mediterranean population. Diabetes Care 2013;36:2850–2856. https:// 85. Miyashita K, Itoh H, Tsujimoto H, Tamura N, Fukunaga Y, Sone M, et al. Natriuretic
doi.org/10.2337/dc12-2337 peptides/cGMP/cGMP-dependent protein kinase cascades promote muscle mito­
63. Celik S, Sadegh MK, Morley M, Roselli C, Ellinor PT, Cappola T, et al. Antisense regu­ chondrial biogenesis and prevent obesity. Diabetes 2009;58:2880–2892. https://doi.
lation of atrial natriuretic peptide expression. JCI Insight 2019;4:e130978. https://doi. org/10.2337/db09-0393
org/10.1172/jci.insight.130978 86. Perez-Ternero C, Aubdool AA, Makwana R, Sanger GJ, Stimson RH, Chan LF, et al.
64. Celik S, Karbalaei-Sadegh M, Rådegran G, Smith JG, Gidlöf O. Functional screening C-type natriuretic peptide is a pivotal regulator of metabolic homeostasis. Proc Natl
identifies microRNA regulators of corin activity and atrial natriuretic peptide biogen­ Acad Sci U S A 2022;119:e2116470119. https://doi.org/10.1073/pnas.2116470119
esis. Mol Cell Biol 2019;39:e00271–19. https://doi.org/10.1128/MCB.00271-19 87. Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PW, et al. Impact of obesity
65. D’Souza SP, Davis M, Baxter GF. Autocrine and paracrine actions of natriuretic pep­ on plasma natriuretic peptide levels. Circulation 2004;109:594–600. https://doi.org/10.
tides in the heart. Pharmacol Ther 2004;101:113–129. https://doi.org/10.1016/j. 1161/01.CIR.0000112582.16683.EA
pharmthera.2003.11.001 88. Undank S, Kaiser J, Sikimic J, Düfer M, Krippeit-Drews P, Drews G. Atrial natriuretic
66. Forte M, Marchitti S, Di Nonno F, Stanzione R, Schirone L, Cotugno M, et al. NPPA/ peptide affects stimulus-secretion coupling of pancreatic β-cells. Diabetes 2017;66:
Atrial natriuretic peptide is an extracellular modulator of autophagy in the heart. 2840–2848. https://doi.org/10.2337/db17-0392
Autophagy 2022. https://doi.org/10.1080/15548627.2022.2115675 89. Hwu CM, Tsai SC, Lau CP, Pu HF, Wang TL, Chiang ST, et al. Increased concentrations
67. Pandey KN. Biology of natriuretic peptides and their receptors. Peptides 2005;26: of atrial and plasma atrial natriuretic peptide in castrated Male rats. Life Sci 1993;52:
901–932. https://doi.org/10.1016/j.peptides.2004.09.024 205–212. https://doi.org/10.1016/0024-3205(93)90141-O

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


68. Forte M, Madonna M, Schiavon S, Valenti V, Versaci F, Zoccai GB, et al. Cardiovascular 90. Clerico A, Giannoni A, Vittorini S, Passino C. Thirty years of the heart as an endocrine
pleiotropic effects of natriuretic peptides. Int J Mol Sci 2019;20:3874. https://doi.org/10. organ: physiological role and clinical utility of cardiac natriuretic hormones. Am J Physiol
3390/ijms20163874 Heart Circ Physiol 2011;301:12–20. https://doi.org/10.1152/ajpheart.00226.2011
69. John SW, Krege JH, Oliver PM, Hagaman JR, Hodgin JB, Pang SC, et al. Genetic de­ 91. Volpe M, Tritto C, De Luca N, Mele AF, Lembo G, Rubattu S, et al. Failure of atrial
creases in atrial natriuretic peptide and salt-sensitive hypertension. Science 1995; natriuretic factor to increase with saline load in patients with dilated cardiomyopathy
267:679–681. https://doi.org/10.1126/science.7839143 and mild heart failure. J Clin Invest 1991;88:1481–1489. https://doi.org/10.1172/
70. Scott NJ, Ellmers LJ, Lainchbury JG, Maeda N, Smithies O, Richards AM, et al. Influence JCI115458
of natriuretic peptide receptor-1 on survival and cardiac hypertrophy during develop­ 92. Hunt PJ, Espiner EA, Nicholls MG, Richards AM, Yandle TG. Differing biological effects
ment. Biochim Biophys Acta 2009;1792:1175–1184. https://doi.org/10.1016/j.bbadis. of equimolar atrial and brain natriuretic peptide infusions in Normal man. J Clin
2009.09.009 Endocrinol Metab 1996;81:3871–3876. https://doi.org/10.1210/jcem.81.11.8923831
71. Rubattu S, Bigatti G, Evangelista A, Lanzani C, Stanzione R, Zagato L, et al. Association 93. Volpe M, Lembo G, De Luca N, Lamenza F, Tritto C, Ricciardelli B, et al. Abnormal
of atrial natriuretic peptide and type a natriuretic peptide receptor gene polymorph­ hormonal and renal responses to saline load in hypertensive patients with parental his­
isms with left ventricular mass in human essential hypertension. J Am Coll Cardiol 2006; tory of cardiovascular accidents. Circulation 1991;84:92–100. https://doi.org/10.1161/
48:499–505. https://doi.org/10.1016/j.jacc.2005.12.081
01.CIR.84.1.92
72. Tamura N, Ogawa Y, Chusho H, Nakamura K, Nakao K, Suda M, et al. Cardiac fibrosis
94. Liang F, O’Rear J, Schellenberger U, Tai L, Lasecki M, Schreiner GF, et al. Evidence for
in mice lacking brain natriuretic peptide. Proc Natl Acad Sci U S A 2000;97:4239–4244.
functional heterogeneity of circulating B-type natriuretic peptide. J Am Coll Cardiol
https://doi.org/10.1073/pnas.070371497
2007;49:1071–1708. https://doi.org/10.1016/j.jacc.2006.10.063
73. Becker JR, Chatterjee S, Robinson TY, Bennett JS, Panáková D, Galindo CL, et al.
95. Nakagawa Y, Nishikimi T, Kuwahara K, Fujishima A, Oka S, Tsutamoto T, et al.
Differential activation of natriuretic peptide receptors modulates cardiomyocyte pro­
MiR30-GALNT1/2 axis-mediated glycosylation contributes to the increased secretion
liferation during development. Development 2014;141:335–345. https://doi.org/10.
of inactive human prohormone for brain natriuretic peptide (proBNP) from failing
1242/dev.100370
hearts. J Am Heart Assoc 2017;6:e003601. https://doi.org/10.1161/JAHA.116.003601
74. Rubattu S, Sciarretta S, Valenti V, Stanzione R, Volpe M. Natriuretic peptides: an up­
96. Supaporn T, Sandberg SM, Borgeson DD, Heublein DM, Luchner A, Wei CM, et al.
date on bioactivity, potential therapeutic use, and implication in cardiovascular dis­
Blunted cGMP response to agonists and enhanced glomerular cyclic 3’,5’-nucleotide
eases. Am J Hypertens 2008;21:733–741. https://doi.org/10.1038/ajh.2008.174
phosphodiesterase activities in experimental congestive heart failure. Kidney Int
75. Sciarretta S, Marchitti S, Bianchi F, Moyes A, Barbato E, Di Castro S, et al. C2238 atrial
1996;50:1718–1725. https://doi.org/10.1038/ki.1996.491
natriuretic peptide molecular variant is associated with endothelial damage and dys­
97. Stasch JP, Becker EM, Alonso-Alija C, Apeler H, Dembowsky K, Feurer A, et al.
function through natriuretic peptide receptor C signaling. Circ Res 2013;112:
NOindependent regulatory site on soluble guanylate cyclase. Nature 2001;410:
1355–1364. https://doi.org/10.1161/CIRCRESAHA.113.301325
212–215. https://doi.org/10.1038/35065611
76. Kuhn M, Völker K, Schwarz K, Carbajo-Lozoya J, Flögel U, Jacoby C, et al. The natri­
98. Gallo G, Rubattu S, Volpe M. Targeting cyclic guanylate monophosphate in resistant
uretic peptide/guanylyl cyclase–a system functions as a stress-responsive regulator
hypertension and heart failure: are sacubitril/valsartan and vericiguat synergistic and ef­
of angiogenesis in mice. J Clin Invest 2009;119:2019–2030. https://doi.org/10.1172/
fective in both conditions? High Blood Press Cardiovasc Prev 2021;28:541–545. https://
JCI37430
77. Špiranec Spes K, Chen W, Krebes L, Völker K, Abeßer M, Eder Negrin P, et al. doi.org/10.1007/s40292-021-00489-z
Heart-microcirculation connection: effects of ANP (atrial natriuretic peptide) on peri­ 99. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, et al. 2021
cytes participate in the acute and chronic regulation of arterial blood pressure. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure.
Hypertension 2020;76:1637–1648. https://doi.org/10.1161/HYPERTENSIONAHA. Eur Heart J 2021;42:3599–3726. https://doi.org/10.1093/eurheartj/ehab368
120.15772 100. Lee KK, Doudesis D, Anwar M, Astengo F, Chenevier-Gobeaux C, Claessens YE, et al.
78. Špiranec K, Chen W, Werner F, Nikolaev VO, Naruke T, Koch F, et al. Endothelial Development and validation of a decision support tool for the diagnosis of acute heart
C-type natriuretic peptide acts on pericytes to regulate microcirculatory flow and failure: systematic review, meta-analysis, and modelling study. BMJ 2022;377:e068424.
blood pressure. Circulation 2018;138:494–508. https://doi.org/10.1161/ https://doi.org/10.1136/bmj-2021-068424
CIRCULATIONAHA.117.033383 101. Barbato E, Rubattu S, Bartunek J, Berni A, Sarno G, Vanderheyden M, et al. Human cor­
79. Nakao K, Osawa K, Yasoda A, Yamanaka S, Fujii T, Kondo E, et al. The local CNP/GC-B onary atherosclerosis modulates cardiac natriuretic peptide release. Atherosclerosis
system in growth plate is responsible for physiological endochondral bone growth. Sci 2009;206:258–264. https://doi.org/10.1016/j.atherosclerosis.2009.01.033
Rep 2015;5:10554. https://doi.org/10.1038/srep10554 102. Liu X, Zheng L, Han J, Song L, Geng H, Liu Y. Joint analysis of D-dimer, N-terminal pro
80. Nakao K, Kuwahara K, Nishikimi T, Nakagawa Y, Kinoshita H, Minami T, et al. B-type natriuretic peptide, and cardiac troponin I on predicting acute pulmonary em­
Endothelium-derived C-type natriuretic peptide contributes to blood pressure regu­ bolism relapse and mortality. Sci Rep 2021;11:14909. https://doi.org/10.1038/s41598-
lation by maintaining endothelial integrity. Hypertension 2017;69:286–296. https://doi. 021-94346-7
org/10.1161/HYPERTENSIONAHA.116.08219 103. Berghaus TM, Kutsch J, Faul C, von Scheidt W, Schwaiblmair M. The association of
81. Moyes AJ, Khambata RS, Villar I, Bubb KJ, Baliga RS, Lumsden NG, et al. Endothelial N-terminal pro-brain-type natriuretic peptide with hemodynamics and functional cap­
C-type natriuretic peptide maintains vascular homeostasis. J Clin Invest 2014;124: acity in therapy-naive precapillary pulmonary hypertension: results from a cohort
4039–4051. https://doi.org/10.1172/JCI74281 study. BMC Pulm Med 2017;17:167. https://doi.org/10.1186/s12890-017-0521-4
82. Moyes AJ, Chu SM, Aubdool AA, Dukinfield MS, Margulies KB, Bedi KC, et al. C-type 104. Benza RL, Miller DP, Gomberg-Maitland M, Frantz RP, Foreman AJ, Coffey CS, et al.
natriuretic peptide co-ordinates cardiac structure and function. Eur Heart J 2020;41: Predicting survival in pulmonary arterial hypertension: insights from the registry to
1006–1020. https://doi.org/10.1093/eurheartj/ehz093 evaluate early and long-term pulmonary arterial hypertension disease management
83. Mascareno E, Beckles D, Dhar-Mascareno M, Siddiqui MA. Enhanced hypertrophy in (REVEAL). Circulation 2010;122:164–172. https://doi.org/10.1161/CIRCULATIONAHA.
ob/ob mice due to an impairment in expression of atrial natriuretic peptide. Vascul 109.898122
Pharmacol 2009;51:198–204. https://doi.org/10.1016/j.vph.2009.06.005 105. Hijazi Z, Lindahl B, Oldgren J, Andersson U, Lindbäck J, Granger CB, et al. Repeated
84. Lafontan M, Moro C, Berlan M, Crampes F, Sengenes C, Galitzky J. Control of lipolysis measurements of cardiac biomarkers in atrial fibrillation and validation of the ABC
by natriuretic peptides and cyclic GMP. Trends Endocrinol Metab 2008;19:130–137. stroke score over time. J Am Heart Assoc 2017;6:e004851. https://doi.org/10.1161/
https://doi.org/10.1016/j.tem.2007.11.006 JAHA.116.004851
654 M. Volpe et al.

106. Gallo G, Bianchi F, Cotugno M, Volpe M, Rubattu S. Natriuretic peptides, cognitive im­ 126. Felker GM, Ahmad T, Anstrom KJ, Adams KF, Cooper LS, Ezekowitz JA, et al. Rationale
pairment and dementia: an intriguing pathogenic link with implications in hypertension. and design of the GUIDE-IT study: guiding evidence based therapy using biomarker in­
J Clin Med 2020;9:2265. https://doi.org/10.3390/jcm9072265 tensified treatment in heart failure. JACC Heart Fail 2014;2:457–465. https://doi.org/10.
107. Barbato E, Bartunek J, Marchitti S, Mangiacapra F, Stanzione R, Delrue L, et al. 1016/j.jchf.2014.05.007
NT-proANP circulating level is a prognostic marker in stable ischemic heart disease. 127. Volpe M, Carnovali M, Mastromarino V. The natriuretic peptides system in the patho­
Int J Cardiol 2012;155:311–312. https://doi.org/10.1016/j.ijcard.2011.11.057 physiology of heart failure: from molecular basis to treatment. Clin Sci (Lond) 2016;130:
108. Stewart RAH, Kirby A, White HD, Marschner SL, West M, Thompson PL, et al. B-type 57–77. https://doi.org/10.1042/CS20150469
natriuretic peptide and long-term cardiovascular mortality in patients with coronary 128. Bayes-Genis A, Barallat J, Richards AM. A test in context: neprilysin: function, inhibition,
heart disease. J Am Heart Assoc 2022;11:e024616. https://doi.org/10.1161/JAHA.121. and biomarker. J Am Coll Cardiol 2016;68:639–653. https://doi.org/10.1016/j.jacc.2016.
024616 04.060
109. Yildirir A, Acikel S, Ertan C, Aydinalp A, Ozin B, Muderrisoglu H. Value of peri- 129. Volpe M, Bauersachs J, Bayés-Genís A, Butler J, Cohen-Solal A, Gallo G, et al. Sacubitril/
procedural B-type natriuretic peptide levels in predicting cardiac events after elective valsartan for the management of heart failure: a perspective viewpoint on current evi­
percutaneous coronary intervention. Acta Cardiol 2008;63:47–52. https://doi.org/10. dence. Int J Cardiol 2021;327:138–145. https://doi.org/10.1016/j.ijcard.2020.11.071
2143/AC.63.1.2025331 130. Rubattu S, Cotugno M, Forte M, Stanzione R, Bianchi F, Madonna M, et al. Effects of
110. Gallo G, Presta V, Volpe M, Rubattu S. Molecular and clinical implications of natriuretic dual angiotensin type 1 receptor/neprilysin inhibition vs. angiotensin type 1 receptor
peptides in aortic valve stenosis. J Mol Cell Cardiol 2019;129:266–271. https://doi.org/ inhibition on target organ injury in the stroke-prone spontaneously hypertensive

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


10.1016/j.yjmcc.2019.03.011 rat. J Hypertens 2018;36:1902–1914. https://doi.org/10.1097/HJH.0000000000001762
111. Chen S, Redfors B, O’Neill BP, Clavel MA, Pibarot P, Elmariah S, et al. Low and elevated 131. Murphy SP, Prescott MF, Camacho A, Iyer SR, Maisel AS, Felker GM, et al. Atrial natri­
B-type natriuretic peptide levels are associated with increased mortality in patients uretic peptide and treatment with sacubitril/valsartan in heart failure with reduced
with preserved ejection fraction undergoing transcatheter aortic valve replacement: ejection fraction. JACC Heart Fail 2021;9:127–136. https://doi.org/10.1016/j.jchf.2020.
an analysis of the PARTNER II trial and registry. Eur Heart J 2020;41:958–969. 09.013
https://doi.org/10.1093/eurheartj/ehz892 132. Omland T, Aakvaag A, Bonarjee VV, Caidahl K, Lie RT, Nilsen DW, et al. Plasma brain
112. Helgeson SA, Imam JS, Moss JE, Hodge DO, Burger CD. Comparison of brain natri­ natriuretic peptide as an indicator of left ventricular systolic function and long-term
uretic peptide levels to simultaneously obtained right heart hemodynamics in stable survival after acute myocardial infarction. Comparison with plasma atrial natriuretic
outpatients with pulmonary arterial hypertension. Diseases 2018;6:33. https://doi. peptide and N-terminal proatrial natriuretic peptide. Circulation 1996;93:
org/10.3390/diseases6020033 1963–1969. https://doi.org/10.1161/01.CIR.93.11.1963
113. Gallo G, Forte M, Stanzione R, Cotugno M, Bianchi F, Marchitti S, et al. Functional role 133. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, et al.
of natriuretic peptides in risk assessment and prognosis of patients with mitral regur­ Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014;
gitation. J Clin Med 2020;9:1348. https://doi.org/10.3390/jcm9051348
371:993–1004. https://doi.org/10.1056/NEJMoa1409077
114. Romaniello A, Rubattu S, Gigante A, Simonelli F, Grimaldi MC, D’Angelo A, et al. Atrial
134. Senni M, Wachter R, Witte KK, Straburzynska-Migaj E, Belohlavek J, Fonseca C, et al.
natriuretic peptide predicts disease progression and digital ulcers development in sys­
Initiation of sacubitril/valsartan shortly after hospitalisation for acutely decompensated
temic sclerosis patients. J Cardiovasc Med 2019;20:771–779. https://doi.org/10.2459/
heart failure in patients with newly diagnosed (de novo) heart failure: a subgroup ana­
JCM.0000000000000852
lysis of the TRANSITION study. Eur J Heart Fail 2020;22:303–312. https://doi.org/10.
115. Romaniello A, Rubattu S, Vaiarello V, Gigante A, Volpe M, Rosato E. Circulating
1002/ejhf.1670
NT-proANP level is a predictor of mortality for systemic sclerosis: a retrospective
135. Berg DD, Samsky MD, Velazquez EJ, Duffy CI, Gurmu Y, Braunwald E, et al. Efficacy and
study of an Italian cohort. Expert Rev Clin Immunol 2021;17:661–666. https://doi.org/
safety of sacubitril/valsartan in high-risk patients in the PIONEER-HF trial. Circ Heart
10.1080/1744666X.2021.1908888
Fail 2021;14:e007034. https://doi.org/10.1161/CIRCHEARTFAILURE.120.007034
116. Takashio S, Takahama H, Nishikimi T, Hayashi T, Nagai-Okatani C, Matsuo A, et al.
136. Senni M, McMurray JJ, Wachter R, McIntyre HF, Reyes A, Majercak I, et al. Initiating sa­
Superiority of proatrial natriuretic peptide in the prognostic power in patients with
cubitril/valsartan (LCZ696) in heart failure: results of TITRATION, a double-blind, ran­
acute decompensated heart failure on hospital admission: comparison with B-type
domized comparison of two uptitration regimens. Eur J Heart Fail 2016;18:1193–1202.
natriuretic peptide and other natriuretic peptide forms. Open Heart 2019;6:
https://doi.org/10.1002/ejhf.548
e001072. https://doi.org/10.1136/openhrt-2019-001072
137. Solomon SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, et al.
117. Bégué C, Mörner S, Brito D, Hengstenberg C, Cleland JGF, Arbustini E, et al.
Angiotensin-neprilysin inhibition in heart failure with preserved ejection fraction. N
Mid-regional proatrial natriuretic peptide for predicting prognosis in hypertrophic car­
Engl J Med 2019;381:1609–1620. https://doi.org/10.1056/NEJMoa1908655
diomyopathy. Heart 2020;106:196–202. https://doi.org/10.1136/heartjnl-2019-
138. Solomon SD, Vaduganathan M, Claggett B L, Packer M, Zile M, Swedberg K, et al.
314826
118. Mueller C, McDonald K, de Boer RA, Maisel A, Cleland JGF, Kozhuharov N, et al. Heart Sacubitril/valsartan across the spectrum of ejection fraction in heart failure.
failure association of the European Society of Cardiology practical guidance on the use Circulation 2020;141:352–361. https://doi.org/10.1161/CIRCULATIONAHA.119.
of natriuretic peptide concentrations. Eur J Heart Fail 2019;21:715–731. https://doi.org/ 044586
10.1002/ejhf.1494 139. Gallo G, Volpe M, Battistoni A, Russo D, Tocci G, Musumeci MB. Sacubitril/valsartan as
119. Messerli FH, Rimoldi SF, Bangalore S. The transition from hypertension to heart failure: a therapeutic tool across the range of heart failure phenotypes and ejection fraction
contemporary update. JACC Heart Fail 2017;5:543–551. https://doi.org/10.1016/j.jchf. Spectrum. Front Physiol 2021;12:652163. https://doi.org/10.3389/fphys.2021.652163
2017.04.012 140. Novartis (2021). Novartis Entresto® Granted Expanded Indication in Chronic Heart
120. Willeit P, Kaptoge S, Welsh P, Butterworth AS, Chowdhury R, Spackman SA, et al. Failure by FDA. Available online at: https://www.novartis.com/news/media-releases/
Natriuretic peptides and integrated risk assessment for cardiovascular disease: an novartis-entresto-granted-expanded-indication-chronic-heart-failure-fda (accessed
individual-participant-data meta-analysis. Lancet Diabetes Endocrinol 2016;4:840–849. 16 February 2021).
https://doi.org/10.1016/S2213-8587(16)30196-6 141. Mann DL, Givertz MM, Vader JM, Starling RC, Shah P, McNulty SE, et al. Effect of treat­
121. Volpe M, Francia P, Tocci G, Rubattu S, Cangianiello S, Elena Rao MA, et al. Prediction ment with sacubitril/valsartan in patients with advanced heart failure and reduced ejec­
of long-term survival in chronic heart failure by multiple biomarker assessment: a tion fraction: a randomized clinical trial. JAMA Cardiol 2022;7:17–25. https://doi.org/10.
15-year prospective follow-up study. Clin Cardiol 2010;33:700–707. https://doi.org/ 1001/jamacardio.2021.4567
10.1002/clc.20813 142. Vodovar N, Séronde MF, Laribi S, Gayat E, Lassus J, Januzzi JL Jr, et al. Elevated plasma
122. Ledwidge M, Gallagher J, Conlon C, Tallon E, O’Connell E, Dawkins I, et al. Natriuretic B-type natriuretic peptide concentrations directly inhibit circulating neprilysin activity
peptide-based screening and collaborative care for heart failure: the STOP-HF rando­ in heart failure. JACC Heart Fail 2015;3:629–636. https://doi.org/10.1016/j.jchf.2015.03.
mized trial. JAMA 2013;310:66–74. https://doi.org/10.1001/jama.2013.7588 011
123. Cannone V, Ledwidge M, Watson C, McKie PM, Burnett JC Jr, McDonald K. STOP-HF 143. Vodovar N, Cohen-Solal A, Logeart D. Could neprilysin be already inhibited by BNP in
Trial: higher endogenous BNP and cardiovascular protection in subjects at risk for the LIFE trial? JAMA Cardiol 2022;7:656–657. https://doi.org/10.1001/jamacardio.2022.
heart failure. JACC Basic Transl Sci 2021;6:497–504. https://doi.org/10.1016/j.jacbts. 0784
2021.05.001 144. Pfeffer MA, Claggett B, Lewis EF, Granger CB, Køber L, Maggioni AP, et al. Angiotensin
124. Huelsmann M, Neuhold S, Resl M, Strunk G, Brath H, Francesconi C, et al. PONTIAC receptor-neprilysin inhibition in acute myocardial infarction. N Engl J Med 2021;385:
(NT-proBNP selected prevention of cardiac events in a population of diabetic patients 1845–1855. https://doi.org/10.1056/NEJMoa2104508
without a history of cardiac disease): a prospective randomized controlled trial. J Am 145. Berwanger O, Pfeffer M, Claggett B, Jering KS, Maggioni AP, Steg PG, et al. Sacubitril/
Coll Cardiol 2013;62:1365–1372. https://doi.org/10.1016/j.jacc.2013.05.069 valsartan versus ramipril for patients with acute myocardial infarction: win-ratio ana­
125. Moe GW, Ezekowitz JA, O’Meara E, Lepage S, Howlett JG, Fremes S, et al. The 2014 lysis of the PARADISE-MI trial. Eur J Heart Fail 2022;24:1918–1927. https://doi.org/
Canadian Cardiovascular Society heart failure management guidelines focus update: 10.1002/ejhf.2663
anemia, biomarkers, and recent therapeutic trial implications. Can J Cardiol 2015;31: 146. Tokudome T, Otani K, Mao Y, Jensen LJ, Arai Y, Miyazaki T, et al. Endothelial natriuretic
3–16. https://doi.org/10.1016/j.cjca.2014.10.022 peptide receptor 1 play crucial role for acute and chronic blood pressure regulation by
Natriuretic peptides in clinical practice 655

atrial natriuretic peptide. Hypertension 2022;79:1409–1422. https://doi.org/10.1161/ a randomised, crossover, double-blind, placebo-controlled trial. Lancet Diabetes
HYPERTENSIONAHA.121.18114 Endocrinol 2013;1:19–27. https://doi.org/10.1016/S2213-8587(13)70029-9
147. Macheret F, Heublein D, Costello-Boerrigter LC, Boerrigter G, McKie P, Bellavia D, 158. Burnett JC Jr, Korinek J. The tumultuous journey of nesiritide: past, present, and future.
et al. Human hypertension is characterized by a lack of activation of the antihyperten­ Circ Heart Fail 2008;1:6–8. https://doi.org/10.1161/CIRCHEARTFAILURE.108.776294
sive cardiac hormones ANP and BNP. J Am Coll Cardiol 2012;60:1558–1565. https://doi. 159. Nogi K, Ueda T, Matsue Y, Nogi M, Ishihara S, Nakada Y, et al. Effect of carperitide on
org/10.1016/j.jacc.2012.05.049 the 1 year prognosis of patients with 5 acute decompensated heart failure. ESC Heart
148. Rubattu S, Gallo G. The natriuretic peptides for hypertension treatment. High Blood Fail 2022;9:1061–1070. https://doi.org/10.1002/ehf2.13770
Press Cardiovasc Prev 2022;29:15–21. https://doi.org/10.1007/s40292-021-00483-5 160. Lee CY, Huntley BK, McCormick DJ, Ichiki T, Sangaralingham SJ, Lisy O, et al.
149. Chen HH, Wan SH, Iyer SR, Cannone V, Sangaralingham SJ, Nuetel J, et al. Cenderitide: structural requirements for the creation of a novel dual particulate gua­
First-in-Human study of MANP: a novel ANP (atrial natriuretic peptide) analog in hu­ nylyl cyclase receptor agonist with renal-enhancing in vivo and ex vivo actions. Eur
man hypertension. Hypertension 2021;78:1859–1867. https://doi.org/10.1161/ Heart J Cardiovasc Pharmacother 2016;2:98–105. https://doi.org/10.1093/ehjcvp/pvv040
HYPERTENSIONAHA.121.17159 161. Chan ML, Qi Y, Larimore K, Cherukuri A, Seid L, Jayaram K, et al. Pharmacokinetics and
150. Cannone V, Burnett JC Jr. Natriuretic peptides and blood pressure homeostasis: im­ exposure-response of vosoritide in children with achondroplasia. Clin Pharmacokinet
plications for MANP, a novel guanylyl cyclase a receptor activator for hypertension. 2022;61:263–280. https://doi.org/10.1007/s40262-021-01059-1
Front Physiol 2022;12:815796. https://doi.org/10.3389/fphys.2021.815796 162. Pan S, Chen HH, Dickey DM, Boerrigter G, Lee C, Kleppe LS, et al. Biodesign of a renal-
151. Hodgson-Zingman DM, Karst ML, Zingman LV, Heublein DM, Darbar D, Herron KJ, protective peptide based on alternative splicing of B-type natriuretic peptide. Proc Natl

Downloaded from https://academic.oup.com/eurheartj/article/44/8/643/6965387 by guest on 18 December 2023


et al. Atrial natriuretic peptide frameshift mutation in familial atrial fibrillation. N Engl Acad Sci U S A 2009;106:11282–11287. https://doi.org/10.1073/pnas.0811851106
J Med 2008;359:158–165. https://doi.org/10.1056/NEJMoa0706300 163. Mezo AR, McDonnell KA, Low SC, Song J, Reidy TJ, Lu Q, et al. Atrial natriuretic
152. Volpe M, Gallo G, Rubattu S. Novel ANP (atrial natriuretic peptide)-based therapy for peptide-fc, ANP-fc, fusion proteins: semisynthesis, in vitro activity and pharmacokinet­
hypertension: the promising role of a disease mechanism targeted approach. ics in rats. Bioconjug Chem 2012;23:518–526. https://doi.org/10.1021/bc200592c
Hypertension 2021;78:1868–1870. https://doi.org/10.1161/HYPERTENSIONAHA. 164. Sangaralingham SJ, Kuhn M, Cannone V, Chen HH, Burnett JC. Natriuretic peptide
121.18264 pathways in heart failure—further therapeutic possibilities. Cardiovasc Res 2022:
153. Altara R, da Silva GJJ, Frisk M, Spelta F, Zouein FA, Louch WE, et al. Cardioprotective cvac125. https://doi.org/10.1093/cvr/cvac125
effects of the novel compound vastiras in a preclinical model of End-organ damage. 165. Ichiki T, Jinno A, Tsuji Y. Natriuretic peptide-based novel therapeutics: long journeys of
Hypertension 2020;75:1195–1204. https://doi.org/10.1161/HYPERTENSIONAHA. drug developments optimized for disease states. Biology (Basel) 2022;11:859. https://
120.14704 doi.org/10.3390/biology11060859
154. Gidlöf O. Toward a new paradigm for targeted natriuretic peptide enhancement in 166. Sangaralingham SJ, Whig K, Peddibhotla S, Kirby RJ, Sessions HE, Maloney PR, et al.
heart failure. Front Physiol 2021;12:650124. https://doi.org/10.3389/fphys.2021.650124 Discovery of small molecule guanylyl cyclase A receptor positive allosteric modulators.
155. Newby DE, McDonagh T, Currie PF, Northridge DB, Boon NA, Dargie HJ. Proc Natl Acad Sci U S A 2021;118:e2109386118. https://doi.org/10.1073/pnas.
Candoxatril improves exercise capacity in patients with chronic heart failure receiving 2109386118
angiotensin converting enzyme inhibition. Eur Heart J 1998;19:1808–1813. https://doi. 167. Preedy MEJ. Cardiac cyclic nucleotide phosphodiesterases: roles and therapeutic po­
org/10.1053/euhj.1998.1201 tential in heart failure. Cardiovasc Drugs Ther 2020;34:401–417. https://doi.org/10.
156. Packer M, Califf RM, Konstam MA, Krum H, McMurray JJ, Rouleau JL, et al. Comparison 1007/s10557-020-06959-1
of omapatrilat and enalapril in patients with chronic heart failure: the omapatrilat ver­ 168. Armstrong PW, Pieske B, Anstrom KJ, Ezekowitz J, Hernandez AF, Butler J, et al.
sus enalapril randomized trial of utility in reducing events (OVERTURE). Circulation Vericiguat in patients with heart failure and reduced ejection fraction. N Engl J Med
2002;106:920–926. https://doi.org/10.1161/01.CIR.0000029801.86489.50 2020;382:1883–1893. https://doi.org/10.1056/NEJMoa1915928
157. Parvanova A, van der Meer IM, Iliev I, Perna A, Gaspari F, Trevisan R, et al. Effect on 169. Schmidt P, Schramm M, Schroder H, Stasch JP. Mechanisms of nitric oxide independent
blood pressure of combined inhibition of endothelin-converting enzyme and neutral activation of soluble guanylyl cyclase. Eur J Pharmacol 2003;468:167–174. https://doi.
endopeptidase with daglutril in patients with type 2 diabetes who have albuminuria: org/10.1016/S0014-2999(03)01674-1

Erratum https://doi.org/10.1093/eurheartj/ehac775
Online publish-ahead-of-print 22 December 2022
.....................................................................................................................................................................................

Erratum to: Transfemoral aortic valve implantation: procedural hospital volume and mortality in Germany
This is an erratum to: Kurt Bestehorn, Maike Bestehorn, Ralf Zahn, Christian Perings, Christoph Stellbrink, Volker Schächinger, Transfemoral
aortic valve implantation: procedural hospital volume and mortality in Germany, European Heart Journal, 2022, ehac698, https://doi.org/
10.1093/eurheartj/ehac698
Upon original publication, Figure 1 was inadvertently omitted from the PDF version of this manuscript.
The Publisher apologizes for this error, which has now been corrected.
© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com

You might also like